Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
J Neurosci ; 41(19): 4187-4201, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33820857

RESUMO

Release of neuropeptides from dense core vesicles (DCVs) is essential for neuromodulation. Compared with the release of small neurotransmitters, much less is known about the mechanisms and proteins contributing to neuropeptide release. By optogenetics, behavioral analysis, electrophysiology, electron microscopy, and live imaging, we show that synapsin SNN-1 is required for cAMP-dependent neuropeptide release in Caenorhabditis elegans hermaphrodite cholinergic motor neurons. In synapsin mutants, behaviors induced by the photoactivated adenylyl cyclase bPAC, which we previously showed to depend on ACh and neuropeptides (Steuer Costa et al., 2017), are altered as in animals with reduced cAMP. Synapsin mutants have slight alterations in synaptic vesicle (SV) distribution; however, a defect in SV mobilization was apparent after channelrhodopsin-based photostimulation. DCVs were largely affected in snn-1 mutants: DCVs were ∼30% reduced in synaptic terminals, and their contents not released following bPAC stimulation. Imaging axonal DCV trafficking, also in genome-engineered mutants in the serine-9 protein kinase A phosphorylation site, showed that synapsin captures DCVs at synapses, making them available for release. SNN-1 colocalized with immobile, captured DCVs. In synapsin deletion mutants, DCVs were more mobile and less likely to be caught at release sites, and in nonphosphorylatable SNN-1B(S9A) mutants, DCVs traffic less and accumulate, likely by enhanced SNN-1 dependent tethering. Our work establishes synapsin as a key mediator of neuropeptide release.SIGNIFICANCE STATEMENT Little is known about mechanisms that regulate how neuropeptide-containing dense core vesicles (DCVs) traffic along the axon, how neuropeptide release is orchestrated, and where it occurs. We found that one of the longest known synaptic proteins, required for the regulation of synaptic vesicles and their storage in nerve terminals, synapsin, is also essential for neuropeptide release. By electrophysiology, imaging, and electron microscopy in Caenorhabditis elegans, we show that synapsin regulates this process by tethering the DCVs to the cytoskeleton in axonal regions where neuropeptides are to be released. Without synapsin, DCVs cannot be captured at the release sites and, consequently, cannot fuse with the membrane, and neuropeptides are not released. We suggest that synapsin fulfills this role also in vertebrates, including humans.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiologia , AMP Cíclico/metabolismo , Neuropeptídeos/metabolismo , Sinapsinas/genética , Sinapsinas/fisiologia , Vesículas Sinápticas/fisiologia , Animais , Animais Geneticamente Modificados , Comportamento Animal , Caenorhabditis elegans , Fenômenos Eletrofisiológicos , Mutação , Optogenética , Estimulação Luminosa , Terminações Pré-Sinápticas , Transmissão Sináptica/genética , Vesículas Sinápticas/genética
2.
Int J Mol Sci ; 21(24)2020 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-33352696

RESUMO

COASY protein-associated neurodegeneration (CoPAN) is a rare but devastating genetic autosomal recessive disorder of inborn error of CoA metabolism, which shares with pantothenate kinase-associated neurodegeneration (PKAN) similar features, such as dystonia, parkinsonian traits, cognitive impairment, axonal neuropathy, and brain iron accumulation. These two disorders are part of the big group of neurodegenerations with brain iron accumulation (NBIA) for which no effective treatment is available at the moment. To date, the lack of a mammalian model, fully recapitulating the human disorder, has prevented the elucidation of pathogenesis and the development of therapeutic approaches. To gain new insights into the mechanisms linking CoA metabolism, iron dyshomeostasis, and neurodegeneration, we generated and characterized the first CoPAN disease mammalian model. Since CoA is a crucial metabolite, constitutive ablation of the Coasy gene is incompatible with life. On the contrary, a conditional neuronal-specific Coasy knock-out mouse model consistently developed a severe early onset neurological phenotype characterized by sensorimotor defects and dystonia-like movements, leading to premature death. For the first time, we highlighted defective brain iron homeostasis, elevation of iron, calcium, and magnesium, together with mitochondrial dysfunction. Surprisingly, total brain CoA levels were unchanged, and no signs of neurodegeneration were present.


Assuntos
Coenzima A Ligases/fisiologia , Hemocromatose/patologia , Ferro/metabolismo , Doenças Mitocondriais/patologia , Transtornos Motores/patologia , Neurodegeneração Associada a Pantotenato-Quinase/complicações , Sinapsinas/fisiologia , Animais , Coenzima A/metabolismo , Feminino , Hemocromatose/etiologia , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Doenças Mitocondriais/etiologia , Doenças Mitocondriais/metabolismo , Transtornos Motores/etiologia , Transtornos Motores/metabolismo
3.
Cereb Cortex ; 30(3): 1393-1406, 2020 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-31504258

RESUMO

Neurotransmitters can be released either synchronously or asynchronously with respect to action potential timing. Synapsins (Syns) are a family of synaptic vesicle (SV) phosphoproteins that assist gamma-aminobutyric acid (GABA) release and allow a physiological excitation/inhibition balance. Consistently, deletion of either or both Syn1 and Syn2 genes is epileptogenic. In this work, we have characterized the effect of SynI knockout (KO) in the regulation of GABA release dynamics. Using patch-clamp recordings in hippocampal slices, we demonstrate that the lack of SynI impairs synchronous GABA release via a reduction of the readily releasable SVs and, in parallel, increases asynchronous GABA release. The effects of SynI deletion on synchronous GABA release were occluded by ω-AgatoxinIVA, indicating the involvement of P/Q-type Ca2+channel-expressing neurons. Using in situ hybridization, we show that SynI is more expressed in parvalbumin (PV) interneurons, characterized by synchronous release, than in cholecystokinin or SOM interneurons, characterized by a more asynchronous release. Optogenetic activation of PV and SOM interneurons revealed a specific reduction of synchronous release in PV/SynIKO interneurons associated with an increased asynchronous release in SOM/SynIKO interneurons. The results demonstrate that SynI is differentially expressed in interneuron subpopulations, where it boosts synchronous and limits asynchronous GABA release.


Assuntos
Interneurônios/fisiologia , Sinapsinas/fisiologia , Transmissão Sináptica , Ácido gama-Aminobutírico/fisiologia , Animais , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Hipocampo/fisiologia , Potenciais Pós-Sinápticos Inibidores , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal , Sinapsinas/genética
4.
eNeuro ; 6(4)2019.
Artigo em Inglês | MEDLINE | ID: mdl-31387877

RESUMO

Synapse formation can be promoted by intense activity. At the Drosophila larval neuromuscular junction (NMJ), new synaptic boutons can grow acutely in response to patterned stimulation. We combined confocal imaging with electron microscopy and tomography to investigate the initial stages of growth and differentiation of new presynaptic boutons at the Drosophila NMJ. We found that the new boutons can form rapidly in intact larva in response to intense crawling activity, and we observed two different patterns of bouton formation and maturation. The first pathway involves the growth of filopodia followed by a formation of boutons that are initially devoid of synaptic vesicles (SVs) but filled with filamentous matrix. The second pathway involves rapid budding of synaptic boutons packed with SVs, and these more mature boutons are sometimes capable of exocytosis/endocytosis. We demonstrated that intense activity predominantly promotes the second pathway, i.e., budding of more mature boutons filled with SVs. We also showed that this pathway depends on synapsin (Syn), a neuronal protein which reversibly associates with SVs and mediates their clustering via a protein kinase A (PKA)-dependent mechanism. Finally, we took advantage of the temperature-sensitive mutant sei to demonstrate that seizure activity can promote very rapid budding of new boutons filled with SVs, and this process occurs at scale of minutes. Altogether, these results demonstrate that intense activity acutely and selectively promotes rapid budding of new relatively mature presynaptic boutons filled with SVs, and that this process is regulated via a PKA/Syn-dependent pathway.


Assuntos
Locomoção , Junção Neuromuscular/fisiologia , Terminações Pré-Sinápticas/fisiologia , Vesículas Sinápticas/fisiologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Drosophila , Proteínas de Drosophila/fisiologia , Feminino , Masculino , Junção Neuromuscular/citologia , Junção Neuromuscular/crescimento & desenvolvimento , Junção Neuromuscular/ultraestrutura , Terminações Pré-Sinápticas/ultraestrutura , Sinapsinas/fisiologia
5.
Mol Metab ; 16: 88-99, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30100244

RESUMO

OBJECTIVE: Insulin resistance is associated with impaired receptor dependent hepatic uptake of triglyceride-rich lipoproteins (TRL), promoting hypertriglyceridemia and atherosclerosis. Next to low-density lipoprotein (LDL) receptor (LDLR) and syndecan-1, the LDLR-related protein 1 (LRP1) stimulated by insulin action contributes to the rapid clearance of TRL in the postprandial state. Here, we investigated the hypothesis that the adaptor protein phosphotyrosine interacting domain-containing protein 1 (PID1) regulates LRP1 function, thereby controlling hepatic endocytosis of postprandial lipoproteins. METHODS: Localization and interaction of PID1 and LRP1 in cultured hepatocytes was studied by confocal microscopy of fluorescent tagged proteins, by indirect immunohistochemistry of endogenous proteins, by GST-based pull down and by immunoprecipitation experiments. The in vivo relevance of PID1 was assessed using whole body as well as liver-specific Pid1-deficient mice on a wild type or Ldlr-deficient (Ldlr-/-) background. Intravital microscopy was used to study LRP1 translocation in the liver. Lipoprotein metabolism was investigated by lipoprotein profiling, gene and protein expression as well as organ-specific uptake of radiolabelled TRL. RESULTS: PID1 co-localized in perinuclear endosomes and was found associated with LRP1 under fasting conditions. We identified the distal NPxY motif of the intracellular C-terminal domain (ICD) of LRP1 as the site critical for the interaction with PID1. Insulin-mediated NPxY-phosphorylation caused the dissociation of PID1 from the ICD, causing LRP1 translocation to the plasma membrane. PID1 deletion resulted in higher LRP1 abundance at the cell surface, higher LDLR protein levels and, paradoxically, reduced total LRP1. The latter can be explained by higher receptor shedding, which we observed in cultured Pid1-deficient hepatocytes. Consistently, PID1 deficiency alone led to increased LDLR-dependent endocytosis of postprandial lipoproteins and lower plasma triglycerides. In contrast, hepatic PID1 deletion on an Ldlr-/- background reduced lipoprotein uptake into liver and caused plasma TRL accumulation. CONCLUSIONS: By acting as an insulin-dependent retention adaptor, PID1 serves as a regulator of LRP1 function controlling the disposal of postprandial lipoproteins. PID1 inhibition provides a novel approach to lower plasma levels of pro-atherogenic TRL remnants by stimulating endocytic function of both LRP1 and LDLR in the liver.


Assuntos
Proteínas de Transporte/metabolismo , Hipertrigliceridemia/metabolismo , Lipoproteínas/metabolismo , Triglicerídeos/metabolismo , Animais , Carcinoma Hepatocelular , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Endocitose/fisiologia , Hepatócitos/metabolismo , Humanos , Hipertrigliceridemia/genética , Insulina/metabolismo , Resistência à Insulina/fisiologia , Lipoproteínas/fisiologia , Fígado/metabolismo , Neoplasias Hepáticas , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Período Pós-Prandial , Receptores de LDL/metabolismo , Sinapsinas/metabolismo , Sinapsinas/fisiologia , Triglicerídeos/fisiologia , Proteínas Supressoras de Tumor/metabolismo
6.
Neurosci Bull ; 34(2): 349-358, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29282612

RESUMO

Synapsins serve as flagships among the presynaptic proteins due to their abundance on synaptic vesicles and contribution to synaptic communication. Several studies have emphasized the importance of this multi-gene family of neuron-specific phosphoproteins in maintaining brain physiology. In the recent times, increasing evidence has established the relevance of alterations in synapsins as a major determinant in many neurological disorders. Here, we give a comprehensive description of the diverse roles of the synapsin family and the underlying molecular mechanisms that contribute to several neurological disorders. These physiologically important roles of synapsins associated with neurological disorders are just beginning to be understood. A detailed understanding of the diversified expression of synapsins may serve to strategize novel therapeutic approaches for these debilitating neurological disorders.


Assuntos
Doenças do Sistema Nervoso Central/fisiopatologia , Sinapsinas/fisiologia , Animais , Humanos
7.
J Neurosci ; 36(36): 9391-406, 2016 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-27605614

RESUMO

UNLABELLED: Interactions between the prefrontal cortex (PFC) and basolateral amygdala (BLA) regulate emotional behaviors. However, a circuit-level understanding of functional connections between these brain regions remains incomplete. The BLA sends prominent glutamatergic projections to the PFC, but the overall influence of these inputs is predominantly inhibitory. Here we combine targeted recordings and optogenetics to examine the synaptic underpinnings of this inhibition in the mouse infralimbic PFC. We find that BLA inputs preferentially target layer 2 corticoamygdala over neighboring corticostriatal neurons. However, these inputs make even stronger connections onto neighboring parvalbumin and somatostatin expressing interneurons. Inhibitory connections from these two populations of interneurons are also much stronger onto corticoamygdala neurons. Consequently, BLA inputs are able to drive robust feedforward inhibition via two parallel interneuron pathways. Moreover, the contributions of these interneurons shift during repetitive activity, due to differences in short-term synaptic dynamics. Thus, parvalbumin interneurons are activated at the start of stimulus trains, whereas somatostatin interneuron activation builds during these trains. Together, these results reveal how the BLA impacts the PFC through a complex interplay of direct excitation and feedforward inhibition. They also highlight the roles of targeted connections onto multiple projection neurons and interneurons in this cortical circuit. Our findings provide a mechanistic understanding for how the BLA can influence the PFC circuit, with important implications for how this circuit participates in the regulation of emotion. SIGNIFICANCE STATEMENT: The prefrontal cortex (PFC) and basolateral amygdala (BLA) interact to control emotional behaviors. Here we show that BLA inputs elicit direct excitation and feedforward inhibition of layer 2 projection neurons in infralimbic PFC. BLA inputs are much stronger at corticoamygdala neurons compared with nearby corticostriatal neurons. However, these inputs are even more powerful at parvalbumin and somatostatin expressing interneurons. BLA inputs thus activate two parallel inhibitory networks, whose contributions change during repetitive activity. Finally, connections from these interneurons are also more powerful at corticoamygdala neurons compared with corticostriatal neurons. Together, our results demonstrate how the BLA predominantly inhibits the PFC via a complex sequence involving multiple cell-type and input-specific connections.


Assuntos
Complexo Nuclear Basolateral da Amígdala/fisiologia , Inibição Neural/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/citologia , Sinapsinas/fisiologia , Potenciais Sinápticos/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Channelrhodopsins , Toxina da Cólera/metabolismo , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neurônios/classificação , Parvalbuminas/genética , Parvalbuminas/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Sinapsinas/genética , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/genética
8.
Curr Psychiatry Rep ; 18(8): 77, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27371030

RESUMO

Schizophrenia is a serious psychiatric illness which is experienced by about 1 % of individuals worldwide and has a debilitating impact on perception, cognition, and social function. Over the years, several models/hypotheses have been developed which link schizophrenia to dysregulations of the dopamine, glutamate, and serotonin receptor pathways. An important segment of these pathways that have been extensively studied for the pathophysiology of schizophrenia is the presynaptic neurotransmitter release mechanism. This set of molecular events is an evolutionarily well-conserved process that involves vesicle recruitment, docking, membrane fusion, and recycling, leading to efficient neurotransmitter delivery at the synapse. Accumulated evidence indicate dysregulation of this mechanism impacting postsynaptic signal transduction via different neurotransmitters in key brain regions implicated in schizophrenia. In recent years, after ground-breaking work that elucidated the operations of this mechanism, research efforts have focused on the alterations in the messenger RNA (mRNA) and protein expression of presynaptic neurotransmitter release molecules in schizophrenia and other neuropsychiatric conditions. In this review article, we present recent evidence from schizophrenia human postmortem studies that key proteins involved in the presynaptic release mechanism are dysregulated in the disorder. We also discuss the potential impact of dysfunctional presynaptic neurotransmitter release on the various neurotransmitter systems implicated in schizophrenia.


Assuntos
Encéfalo/fisiopatologia , Esquizofrenia/fisiopatologia , Psicologia do Esquizofrênico , Vesículas Sinápticas/fisiologia , Animais , Encéfalo/patologia , Humanos , Proteínas Munc18/fisiologia , Neurotransmissores/metabolismo , Proteínas Qa-SNARE/fisiologia , Proteínas R-SNARE/fisiologia , RNA Mensageiro/genética , Receptores Pré-Sinápticos/fisiologia , Proteínas SNARE/fisiologia , Esquizofrenia/patologia , Transdução de Sinais/fisiologia , Sinapsinas/fisiologia , Vesículas Sinápticas/genética , Sinaptofisina/fisiologia , Proteína 25 Associada a Sinaptossoma/fisiologia
9.
Cell Stem Cell ; 18(1): 134-43, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26549107

RESUMO

Capturing the full potential of human pluripotent stem cell (PSC)-derived neurons in disease modeling and regenerative medicine requires analysis in complex functional systems. Here we establish optogenetic control in human PSC-derived spinal motorneurons and show that co-culture of these cells with human myoblast-derived skeletal muscle builds a functional all-human neuromuscular junction that can be triggered to twitch upon light stimulation. To model neuromuscular disease we incubated these co-cultures with IgG from myasthenia gravis patients and active complement. Myasthenia gravis is an autoimmune disorder that selectively targets neuromuscular junctions. We saw a reversible reduction in the amplitude of muscle contractions, representing a surrogate marker for the characteristic loss of muscle strength seen in this disease. The ability to recapitulate key aspects of disease pathology and its symptomatic treatment suggests that this neuromuscular junction assay has significant potential for modeling of neuromuscular disease and regeneration.


Assuntos
Células-Tronco Embrionárias/citologia , Neurônios Motores/patologia , Miastenia Gravis/imunologia , Doenças Neuromusculares/fisiopatologia , Junção Neuromuscular/fisiopatologia , Optogenética/métodos , Autoimunidade , Técnicas de Cocultura , Proteínas do Sistema Complemento , Humanos , Imunoglobulina G/química , Imuno-Histoquímica , Luz , Músculo Esquelético/fisiologia , Músculos/fisiologia , Miastenia Gravis/fisiopatologia , Mioblastos/citologia , Células-Tronco Pluripotentes/citologia , Regeneração , Medula Espinal/patologia , Sinapsinas/metabolismo , Sinapsinas/fisiologia
10.
J Physiol ; 593(19): 4499-510, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26256545

RESUMO

KEY POINTS: The synaptic vesicle associated proteins synapsin I and synapsin II have important functions in synaptic short-term plasticity. We investigated their functions in cortical facilitatory feedback to neurons in dorsal lateral geniculate nucleus (dLGN), feedback that has important functions in state-dependent regulation of thalamic transmission of visual input to cortex. We compared results from normal wild-type (WT) mice and synapsin knockout (KO) mice in several types of synaptic plasticity, and found clear differences between the responses of neurons in the synapsin I KO and the WT, but no significant differences between the synapsin II KO and the WT. These results are in contrast to the important role of synapsin II previously demonstrated in similar types of synaptic plasticity in other brain regions, indicating that the synapsins can have different roles in similar types of STP in different parts of the brain. ABSTRACT: The synaptic vesicle associated proteins synapsin I (SynI) and synapsin II (SynII) have important functions in several types of synaptic short-term plasticity in the brain, but their separate functions in different types of synapses are not well known. We investigated possible distinct functions of the two synapsins in synaptic short-term plasticity at corticothalamic synapses on relay neurons in the dorsal lateral geniculate nucleus. These synapses provide excitatory feedback from visual cortex to the relay cells, feedback that can facilitate transmission of signals from retina to cortex. We compared results from normal wild-type (WT), SynI knockout (KO) and SynII KO mice, in three types of synaptic plasticity mainly linked to presynaptic mechanism. In SynI KO mice, paired-pulse stimulation elicited increased facilitation at short interpulse intervals compared to the WT. Pulse-train stimulation elicited weaker facilitation than in the WT, and also post-tetanic potentiation was weaker in SynI KO than in the WT. Between SynII KO and the WT we found no significant differences. Thus, SynI has important functions in these types of synaptic plasticity at corticothalamic synapses. Interestingly, our data are in contrast to the important role of SynII previously shown for sustained synaptic transmission during intense stimulation in excitatory synapses in other parts of the brain, and our results suggest that SynI and SynII may have different roles in similar types of STP in different parts of the brain.


Assuntos
Corpos Geniculados/fisiologia , Plasticidade Neuronal/fisiologia , Sinapsinas/fisiologia , Córtex Visual/fisiologia , Animais , Feminino , Técnicas In Vitro , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sinapsinas/genética
11.
J Neurosci ; 35(19): 7487-502, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25972175

RESUMO

Adverse life events can induce two kinds of memory with opposite valence, dependent on timing: "negative" memories for stimuli preceding them and "positive" memories for stimuli experienced at the moment of "relief." Such punishment memory and relief memory are found in insects, rats, and man. For example, fruit flies (Drosophila melanogaster) avoid an odor after odor-shock training ("forward conditioning" of the odor), whereas after shock-odor training ("backward conditioning" of the odor) they approach it. Do these timing-dependent associative processes share molecular determinants? We focus on the role of Synapsin, a conserved presynaptic phosphoprotein regulating the balance between the reserve pool and the readily releasable pool of synaptic vesicles. We find that a lack of Synapsin leaves task-relevant sensory and motor faculties unaffected. In contrast, both punishment memory and relief memory scores are reduced. These defects reflect a true lessening of associative memory strength, as distortions in nonassociative processing (e.g., susceptibility to handling, adaptation, habituation, sensitization), discrimination ability, and changes in the time course of coincidence detection can be ruled out as alternative explanations. Reductions in punishment- and relief-memory strength are also observed upon an RNAi-mediated knock-down of Synapsin, and are rescued both by acutely restoring Synapsin and by locally restoring it in the mushroom bodies of mutant flies. Thus, both punishment memory and relief memory require the Synapsin protein and in this sense share genetic and molecular determinants. We note that corresponding molecular commonalities between punishment memory and relief memory in humans would constrain pharmacological attempts to selectively interfere with excessive associative punishment memories, e.g., after traumatic experiences.


Assuntos
Aprendizagem por Associação/fisiologia , Aprendizagem da Esquiva/fisiologia , Encéfalo/metabolismo , Memória/fisiologia , Punição , Sinapsinas/fisiologia , Fatores Etários , Animais , Animais Geneticamente Modificados , Encéfalo/citologia , Encéfalo/fisiologia , Discriminação Psicológica , Proteínas de Drosophila/genética , Drosophila melanogaster , Eletrochoque/efeitos adversos , Feminino , Masculino , Mutação/genética , Odorantes , Fosforilação , Interferência de RNA/fisiologia , Sinapsinas/genética
12.
Psychoneuroendocrinology ; 52: 43-58, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25459892

RESUMO

Both the molecular co-chaperone FKBP51 and the presynaptic vesicle protein synapsin (alternatively spliced from SYN1-3) are intensively discussed players in the still insufficiently explored pathobiology of psychiatric disorders such as major depression, schizophrenia and posttraumatic stress disorder (PTSD). To address their still unknown interaction, we compared the expression levels of synapsin and five other neurostructural and HPA axis related marker proteins in the prefrontal cortex (PFC) and the hippocampus of restrained-stressed and unstressed Fkbp5 knockout mice and corresponding wild-type littermates. In addition, we compared and correlated the gene expression levels of SYN1, SYN2 and FKBP5 in three different online datasets comprising expression data of human healthy subjects as well as of predominantly medicated patients with different psychiatric disorders. In summary, we found that Fkbp5 deletion, which we previously demonstrated to improve stress-coping behavior in mice, prevents the stress-induced decline in prefrontal cortical (pc), but not in hippocampal synapsin expression. Accordingly, pc, but not hippocampal, synapsin protein levels correlated positively with a more active mouse stress coping behavior. Searching for an underlying mechanism, we found evidence that deletion of Fkbp5 might prevent stress-induced pc synapsin loss, at least in part, through improvement of pc Akt kinase activity. These results, together with our finding that FKBP5 and SYN1 mRNA levels were regulated in opposite directions in the PFC of schizophrenic patients, who are known for exhibiting an altered stress-coping behavior, provide the first evidence of a role for pc synapsin in FKBP51 modulation of stress responsiveness. This role might extend to other tissues, as we found FKBP5 and SYN1 levels to correlate inversely not only in human PFC samples but also in other expression sites. The main limitation of this study is the small number of individuals included in the correlation analyses. Future studies will have to verify the here-postulated role of the FKBP51-Akt kinase-synapsin pathway in stress responsiveness.


Assuntos
Adaptação Psicológica/fisiologia , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/metabolismo , Sinapsinas/fisiologia , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Comportamento Animal/fisiologia , Expressão Gênica , Humanos , Transtornos Mentais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro
13.
Cereb Cortex ; 25(10): 3356-68, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24962993

RESUMO

Idiopathic epilepsies have frequently been linked to mutations in voltage-gated channels (channelopathies); recently, mutations in several genes encoding presynaptic proteins have been shown to cause epilepsy in humans and mice, indicating that epilepsy can also be considered a synaptopathy. However, the functional mechanisms by which presynaptic dysfunctions lead to hyperexcitability and seizures are not well understood. We show that deletion of synapsin II (Syn II), a presynaptic protein contributing to epilepsy predisposition in humans, leads to a loss of tonic inhibition in mouse hippocampal slices due to a dramatic decrease in presynaptic asynchronous GABA release. We also show that the asynchronous GABA release reduces postsynaptic cell firing, and the parallel impairment of asynchronous GABA release and tonic inhibition results in an increased excitability at both single-neuron and network levels. Restoring tonic inhibition with THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol; gaboxadol), a selective agonist of δ subunit-containing GABAA receptors, fully rescues the SynII(-/-) epileptic phenotype both ex vivo and in vivo. The results demonstrate a causal relationship between the dynamics of GABA release and the generation of tonic inhibition, and identify a novel mechanism of epileptogenesis generated by dysfunctions in the dynamics of release that can be effectively targeted by novel antiepileptic strategies.


Assuntos
Hipocampo/fisiologia , Inibição Neural , Neurônios/fisiologia , Sinapsinas/fisiologia , Ácido gama-Aminobutírico/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Potenciais Pós-Sinápticos Excitadores , Agonistas GABAérgicos/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Potenciais Pós-Sinápticos Inibidores , Isoxazóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Convulsões/fisiopatologia , Sinapsinas/genética
14.
DNA Cell Biol ; 32(8): 430-4, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23768104

RESUMO

It was aimed to investigate the association of the synapsin III gene -196 G> A and -631 C>G polymorphisms that takes place in an encoding presynaptic protein, with adult attention deficit hyperactivity disorder (ADHD). One hundred thirty-nine patients having adult ADHD and 106 controls were included in the study. DNA samples were extracted from whole blood and genetic analyses were performed. A significant difference was determined between ADHD and synapsin III gene -631 C>G polymorphism compared to the control group. No significant difference was determined between ADHD and synapsin III gene -196 G>A polymorphism. These polymorphisms were found not to be associated with subtypes of ADHD. It is supposed that synaptic protein genes together with dopaminergic genes might have roles in the etiology of ADHD.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/genética , Sinapsinas/genética , Adulto , Idade de Início , Alelos , Transtorno do Deficit de Atenção com Hiperatividade/epidemiologia , Estudos de Casos e Controles , Feminino , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Masculino , Polimorfismo de Nucleotídeo Único/fisiologia , Sinapsinas/fisiologia , Adulto Jovem
15.
J Exp Biol ; 216(Pt 6): 939-51, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23447663

RESUMO

Drosophila larvae are focused on feeding and have few neurons. Within these bounds, however, there still are behavioural degrees of freedom. This review is devoted to what these elements of flexibility are, and how they come about. Regarding odour-food associative learning, the emerging working hypothesis is that when a mushroom body neuron is activated as a part of an odour-specific set of mushroom body neurons, and coincidently receives a reinforcement signal carried by aminergic neurons, the AC-cAMP-PKA cascade is triggered. One substrate of this cascade is Synapsin, and therefore this review features a general and comparative discussion of Synapsin function. Phosphorylation of Synapsin ensures an alteration of synaptic strength between this mushroom body neuron and its target neuron(s). If the trained odour is encountered again, the pattern of mushroom body neurons coding this odour is activated, such that their modified output now allows conditioned behaviour. However, such an activated memory trace does not automatically cause conditioned behaviour. Rather, in a process that remains off-line from behaviour, the larvae compare the value of the testing situation (based on gustatory input) with the value of the odour-activated memory trace (based on mushroom body output). The circuit towards appetitive conditioned behaviour is closed only if the memory trace suggests that tracking down the learned odour will lead to a place better than the current one. It is this expectation of a positive outcome that is the immediate cause of appetitive conditioned behaviour. Such conditioned search for reward corresponds to a view of aversive conditioned behaviour as conditioned escape from punishment, which is enabled only if there is something to escape from - much in the same way as we only search for things that are not there, and run for the emergency exit only when there is an emergency. One may now ask whether beyond 'value' additional information about reinforcement is contained in the memory trace, such as information about the kind and intensity of the reinforcer used. The Drosophila larva may allow us to develop satisfyingly detailed accounts of such mnemonic richness - if it exists.


Assuntos
Comportamento Apetitivo/fisiologia , Aprendizagem por Associação/fisiologia , Drosophila melanogaster/fisiologia , Memória/fisiologia , Modelos Biológicos , Olfato/fisiologia , Sinapsinas/metabolismo , Paladar/fisiologia , Animais , Larva/fisiologia , Corpos Pedunculados/citologia , Corpos Pedunculados/fisiologia , Neurônios/fisiologia , Fosforilação , Sinapsinas/fisiologia
16.
Brain Res ; 1486: 53-61, 2012 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-23022567

RESUMO

Stroke is a leading cause of morbidity and mortality worldwide. Recovery of motor function after stroke can be modified by post-injury experience, but most of surviving patients exhibit persistence of the motor dysfunctions even after rehabilitative therapy. In this study we investigated if skilled and unskilled training induce different motor recovery and brain plasticity after experimental focal ischemia. We tested this hypothesis by evaluating the motor skill relearning and the immunocontent of Synapsin-I, PSD-95 and GFAP (pre and post-synaptic elements, as well as surrounding astroglia) in sensorimotor cortex of both hemispheres 6 weeks after endothelin-1-induced focal brain ischemia in rats. Synapsin-I and PSD-95 levels were increased by skilled training in ischemic sensorimotor cortex. The content of GFAP was augmented as a result of focal brain ischemia in ischemic sensorimotor cortex and that was not modified by rehabilitation training. Unexpectedly, animals remained permanently impaired at the end of motor/functional evaluations. Significant modifications in protein expression were not observed in undamaged sensorimotor cortex. We conclude that skilled motor activity can positively affect brain plasticity after focal ischemia despite of no functional improvement in conditions here tested.


Assuntos
Isquemia Encefálica/reabilitação , Aprendizagem/fisiologia , Córtex Motor/fisiologia , Destreza Motora/fisiologia , Plasticidade Neuronal/fisiologia , Recuperação de Função Fisiológica/fisiologia , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Masculino , Córtex Motor/metabolismo , Desempenho Psicomotor/fisiologia , Ratos , Ratos Wistar , Sinapsinas/biossíntese , Sinapsinas/fisiologia
17.
J Neurosci ; 32(35): 12214-27, 2012 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-22933803

RESUMO

The precise subcellular organization of synaptic vesicles (SVs) at presynaptic sites allows for rapid and spatially restricted exocytotic release of neurotransmitter. The synapsins (Syns) are a family of presynaptic proteins that control the availability of SVs for exocytosis by reversibly tethering them to each other and to the actin cytoskeleton in a phosphorylation-dependent manner. Syn ablation leads to reduction in the density of SV proteins in nerve terminals and increased synaptic fatigue under high-frequency stimulation, accompanied by the development of an epileptic phenotype. We analyzed cultured neurons from wild-type and Syn I,II,III(-/-) triple knock-out (TKO) mice and found that SVs were severely dispersed in the absence of Syns. Vesicle dispersion did not affect the readily releasable pool of SVs, whereas the total number of SVs was considerably reduced at synapses of TKO mice. Interestingly, dispersion apparently involved exocytosis-competent SVs as well; it was not affected by stimulation but was reversed by chronic neuronal activity blockade. Altogether, these findings indicate that Syns are essential to maintain the dynamic structural organization of synapses and the size of the reserve pool of SVs during intense SV recycling, whereas an additional Syn-independent mechanism, whose molecular substrate remains to be clarified, targets SVs to synaptic boutons at rest and might be outpaced by activity.


Assuntos
Sinapsinas/deficiência , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/fisiologia , Vesículas Sinápticas/ultraestrutura , Animais , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Sinapsinas/genética , Sinapsinas/fisiologia , Transmissão Sináptica/genética
18.
Neuroscience ; 215: 1-16, 2012 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-22542674

RESUMO

Neuroligins are a family of cell adhesion molecules critical in establishing proper central nervous system connectivity; disruption of neuroligin signaling in vivo precipitates a broad range of cognitive deficits. Despite considerable recent progress, the specific synaptic function of neuroligin-1 (NL1) remains unclear. A current model proposes that NL1 acts exclusively to mature pre-existent synaptic connections in an activity-dependent manner. A second element of this activity-dependent maturation model is that an alternate molecule acts upstream of NL1 to initiate synaptic connections. SynCAM1 (SC1) is hypothesized to function in this capacity, though several uncertainties remain regarding SC1 function. Using overexpression and chronic pharmacological blockade of synaptic activity, we now demonstrate that NL1 is capable of robustly recruiting synapsin-positive terminals independent of synaptic maturation and activity in 2-week old primary hippocampal neuronal cultures. We further report that neither SC1 overexpression nor knockdown of endogenous SC1 impacts synapsin punctum densities, suggesting that SC1 is not a limiting factor of synapse initiation in maturing hippocampal neurons in vitro. Consistent with these findings, we observed profoundly greater recruitment of synapsin-positive presynaptic terminals by NL1 than SC1 in a mixed-culture assay of artificial synaptogenesis between primary neurons and heterologous cells. Collectively, our results contend multiple aspects of the proposed model of NL1 and SC1 function and motivate an alternative model whereby SC1 may mature synaptic connections forged by NL1. Supporting this model, we present evidence that combined NL1 and SC1 overexpression triggers excitotoxic neurodegeneration through SC1 signaling at synaptic connections initiated by NL1.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Regulação da Expressão Gênica/fisiologia , Hipocampo/citologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Proteínas Repressoras/metabolismo , Sinapses/fisiologia , Sinapsinas/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , Análise de Variância , Animais , Biofísica , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Estimulação Elétrica , Embrião de Mamíferos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas Repressoras/genética , Sinapsinas/efeitos dos fármacos , Transfecção
19.
PLoS One ; 7(2): e32680, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384280

RESUMO

Bipolar disorder (BD) is a debilitating psychiatric condition with a prevalence of 1-2% in the general population that is characterized by severe episodic shifts in mood ranging from depressive to manic episodes. One of the most common treatments is lithium (Li), with successful response in 30-60% of patients. Synapsin II (SYN2) is a neuronal phosphoprotein that we have previously identified as a possible candidate gene for the etiology of BD and/or response to Li treatment in a genome-wide linkage study focusing on BD patients characterized for excellent response to Li prophylaxis. In the present study we investigated the role of this gene in BD, particularly as it pertains to Li treatment. We investigated the effect of lithium treatment on the expression of SYN2 in lymphoblastoid cell lines from patients characterized as excellent Li-responders, non-responders, as well as non-psychiatric controls. Finally, we sought to determine if Li has a cell-type-specific effect on gene expression in neuronal-derived cell lines. In both in vitro models, we found SYN2 to be modulated by the presence of Li. By focusing on Li-responsive BD we have identified a potential mechanism for Li response in some patients.


Assuntos
Antipsicóticos/uso terapêutico , Transtorno Bipolar/tratamento farmacológico , Lítio/uso terapêutico , Sinapsinas/fisiologia , Adulto , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Estudo de Associação Genômica Ampla , Células HEK293 , Humanos , Linfócitos/citologia , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Sinapsinas/química
20.
J Neuroimmunol ; 242(1-2): 16-25, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22138356

RESUMO

We previously found that the preventive oral administration of a hybrid consisting of the C domain of synapsin and the B subunit of E. coli heat-labile enterotoxin (LTBSC) efficiently suppresses experimental autoimmune encephalomyelitis (EAE) development in rats. We investigated the effect of LTBSC on cytokine expression and on regulatory T (Treg) cells in rats with myelin induced EAE. LTBSC treatment increased the frequency of CD4(+)FoxP3(+) Treg cells in lymph nodes prior to challenge and in the EAE acute stage. LTBSC also up-regulated the expression of anti-inflammatory Th2/Th3 cytokines and diminished myelin basic protein-specific Th1 and Th17 cell responses in lymph nodes. CD4(+)CD25(+) Treg cells from LTBSC treated rats showed stronger suppressive properties than Treg cells from controls in vitro. Our observations indicate that LTBSC is a useful agent for modulating the autoimmune responses in EAE.


Assuntos
Toxinas Bacterianas/administração & dosagem , Citocinas/biossíntese , Encefalomielite Autoimune Experimental/imunologia , Enterotoxinas/administração & dosagem , Proteínas de Escherichia coli/administração & dosagem , Vacinas contra Escherichia coli/administração & dosagem , Ativação Linfocitária/imunologia , Fragmentos de Peptídeos/fisiologia , Sinapsinas/fisiologia , Linfócitos T Reguladores/imunologia , Animais , Bovinos , Células Cultivadas , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Encefalomielite Autoimune Experimental/microbiologia , Encefalomielite Autoimune Experimental/patologia , Escherichia coli Enterotoxigênica/genética , Escherichia coli Enterotoxigênica/imunologia , Vacinas contra Escherichia coli/genética , Vacinas contra Escherichia coli/imunologia , Feminino , Mediadores da Inflamação/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Linfonodos/microbiologia , Ativação Linfocitária/genética , Masculino , Proteína Básica da Mielina/antagonistas & inibidores , Fragmentos de Peptídeos/genética , Distribuição Aleatória , Ratos , Ratos Wistar , Sinapsinas/genética , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/microbiologia , Regulação para Cima/genética , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA